51
|
Cavalli G, Cenci S. Autophagy and Protein Secretion. J Mol Biol 2020; 432:2525-2545. [PMID: 31972172 DOI: 10.1016/j.jmb.2020.01.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 12/13/2022]
Abstract
Autophagy - conventional for macroautophagy - is a major recycling strategy that ensures cellular homeostasis through the selective engulfment of cytoplasmic supramolecular cargos in double membrane vesicles and their rapid dispatch to the lysosome for digestion. As autophagy operates in the cytoplasm, its interference with secretory proteins, that is, proteins destined to the plasma membrane or the extracellular space, generally synthesized and routed within the endoplasmic reticulum (ER), has been relatively overlooked in the past. However, mounting evidence reveals that autophagy in fact heavily regulates protein secretion through diverse mechanisms. First, autophagy is closely involved in the unconventional secretion of leaderless proteins, a pool of proteins destined extracellularly, but lacking an ER-targeted leader sequence, and thus manufactured in the cytosol. Autophagy-related (ATG) genes now appear instrumental to the underlying pathways, hence the recently coined concept of secretory autophagy, or better ATG gene-dependent secretion. Indeed, ATG genes regulate unconventional protein secretion at multiple levels, ranging from intracellular inflammatory signaling, for example, through the control of mitochondrial health and inflammasome activity, to trafficking of leaderless proteins. Moreover, perhaps less expectedly, autophagy also participates in the control of conventional secretion, intersecting the secretory apparatus at multiple points, though with surprising differences among professional secretory cell types that disclose remarkable and unpredicted specificity. This review synopsizes the multiple mechanisms whereby autophagy interfaces with conventional and unconventional protein secretory pathways and discusses the relative teleology. Altogether, the diverse controls exerted on protein secretion broaden and deepen the homeostatic significance of autophagy within the cell.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Simone Cenci
- Vita-Salute San Raffaele University, Milano, Italy; Unit of Age Related Diseases, Division of Genetics and Cell Biology, Ospedale San Raffaele, Milano, Italy.
| |
Collapse
|
52
|
Abstract
Thiol isomerases are oxidoreductases that mediate disulphide bond formation in nascent proteins of the endoplasmic reticulum to ensure their structural integrity. In addition to its role in protein folding, thiol isomerases can modify allosteric disulphide bonds in both intracellular and extracellular proteins, thereby controlling protein function. The process of disulphide bond formation and cleavage is strictly regulated and responsive to redox conditions. Understanding disulphide bond regulation under different redox environments is critical to understanding physiological and pathological processes related to disulphide bond chemistry. Here we describe protocols for the measurement of disulphide bond modulation by thiol isomerases, including reductase and denitrosylase assays. These methods can be applied to study recombinant thiol isomerases and thiol isomerases in cellular settings.
Collapse
Affiliation(s)
- Roelof H Bekendam
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
53
|
Zai JA, Khan MR, Mughal ZUN, Batool R, Naz I, Maryam S, Zahra Z. Methanol extract of Iphiona aucheri ameliorates CCl 4 induced hepatic injuries by regulation of genes in rats. Toxicol Res (Camb) 2019; 8:815-832. [PMID: 34055308 PMCID: PMC8142630 DOI: 10.1039/c9tx00157c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
We have investigated the protective potential of methanol extract of Iphiona aucheri (IAM) on the expression of endoplasmic reticulum (ER) stress associated genes and inflammatory genes on carbon tetrachloride (CCl4) induced hepatic toxicity in rats. Hepatic damage markers: aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP) and bilirubin were elevated while the content of antioxidants: catalase (CAT), superoxide dismutase (SOD), peroxidase (POD) and reduced glutathione (GSH) were decreased significantly (p < 0.05) in CCl4 treated rats as compared to the control group. The CCl4 intoxication induced a higher expression of glucose-regulated protein 78 kDa (GRP78), X-box-binding protein 1 total (XBP1t), spliced X-box-binding protein 1 (XBP1s), unspliced X-box-binding protein 1 (XBP1u), C/EBP homologous protein (CHOP) and genes involved in inflammation and fibrosis: tumor necrosis factor alpha (TNF-α), transforming growth factor-beta (TGF-β), mothers against DPP homolog 3 (SMAD3), alpha skeletal muscle actin (αSMA) and collagen type I alpha 1 chain (COL1A1). The intoxicated rats showed a low expression of the glutamate-cysteine ligase catalytic subunit (GCLC), protein disulfide isomerase (PDI) and nuclear factor (erythroid-derived 2) like-2 (Nrf2). The administration of IAM to intoxicated rats restored the expression of ER stress, inflammatory, fibrosis and antioxidant genes in a dose dependent manner. Our results indicated that IAM can impede the ER stress and inflammatory genes and it could be a complementary and alternative therapeutic agent for oxidative stress associated disorders.
Collapse
Affiliation(s)
- Jawaid Ahmed Zai
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Muhammad Rashid Khan
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Zaib Un Nisa Mughal
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Riffat Batool
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Irum Naz
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Sonia Maryam
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| | - Zartash Zahra
- Department of Biochemistry , Faculty of Biological Sciences , Quaid-i-azam University Islamabad , Islamabad , Pakistan . ; ; ; ; ; ;
| |
Collapse
|
54
|
Mechanistic Connections between Endoplasmic Reticulum (ER) Redox Control and Mitochondrial Metabolism. Cells 2019; 8:cells8091071. [PMID: 31547228 PMCID: PMC6769559 DOI: 10.3390/cells8091071] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/21/2022] Open
Abstract
The past decade has seen the emergence of endoplasmic reticulum (ER) chaperones as key determinants of contact formation between mitochondria and the ER on the mitochondria-associated membrane (MAM). Despite the known roles of ER–mitochondria tethering factors like PACS-2 and mitofusin-2, it is not yet entirely clear how they mechanistically interact with the ER environment to determine mitochondrial metabolism. In this article, we review the mechanisms used to communicate ER redox and folding conditions to the mitochondria, presumably with the goal of controlling mitochondrial metabolism at the Krebs cycle and at the electron transport chain, leading to oxidative phosphorylation (OXPHOS). To achieve this goal, redox nanodomains in the ER and the interorganellar cleft influence the activities of ER chaperones and Ca2+-handling proteins to signal to mitochondria. This mechanism, based on ER chaperones like calnexin and ER oxidoreductases like Ero1α, controls reactive oxygen production within the ER, which can chemically modify the proteins controlling ER–mitochondria tethering, or mitochondrial membrane dynamics. It can also lead to the expression of apoptotic or metabolic transcription factors. The link between mitochondrial metabolism and ER homeostasis is evident from the specific functions of mitochondria–ER contact site (MERC)-localized Ire1 and PERK. These functions allow these two transmembrane proteins to act as mitochondria-preserving guardians, a function that is apparently unrelated to their functions in the unfolded protein response (UPR). In scenarios where ER stress cannot be resolved via the activation of mitochondrial OXPHOS, MAM-localized autophagosome formation acts to remove defective portions of the ER. ER chaperones such as calnexin are again critical regulators of this MERC readout.
Collapse
|
55
|
The molecular basis of chaperone-mediated interleukin 23 assembly control. Nat Commun 2019; 10:4121. [PMID: 31511508 PMCID: PMC6739322 DOI: 10.1038/s41467-019-12006-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/14/2019] [Indexed: 01/20/2023] Open
Abstract
The functionality of most secreted proteins depends on their assembly into a defined quaternary structure. Despite this, it remains unclear how cells discriminate unassembled proteins en route to the native state from misfolded ones that need to be degraded. Here we show how chaperones can regulate and control assembly of heterodimeric proteins, using interleukin 23 (IL-23) as a model. We find that the IL-23 α-subunit remains partially unstructured until assembly with its β-subunit occurs and identify a major site of incomplete folding. Incomplete folding is recognized by different chaperones along the secretory pathway, realizing reliable assembly control by sequential checkpoints. Structural optimization of the chaperone recognition site allows it to bypass quality control checkpoints and provides a secretion-competent IL-23α subunit, which can still form functional heterodimeric IL-23. Thus, locally-restricted incomplete folding within single-domain proteins can be used to regulate and control their assembly. It is unclear how unassembled secretory pathway proteins are discriminated from misfolded ones. Here the authors combine biophysical and cellular experiments to study the folding of heterodimeric interleukin 23 and describe how ER chaperones recognize unassembled proteins and aid their assembly into protein complexes while preventing the premature degradation of unassembled units.
Collapse
|
56
|
Guyette J, Evangelista B, Tatulian SA, Teter K. Stability and Conformational Resilience of Protein Disulfide Isomerase. Biochemistry 2019; 58:3572-3584. [PMID: 31393106 PMCID: PMC6876119 DOI: 10.1021/acs.biochem.9b00405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein disulfide isomerase (PDI) is a redox-dependent protein with oxidoreductase and chaperone activities. It is a U-shaped protein with an abb'xa' structural organization in which the a and a' domains have CGHC active sites, the b and b' domains are involved with substrate binding, and x is a flexible linker. PDI exhibits substantial flexibility and undergoes cycles of unfolding and refolding in its interaction with cholera toxin, suggesting PDI can regain a folded, functional conformation after exposure to stress conditions. To determine whether this unfolding-refolding cycle is a substrate-induced process or an intrinsic physical property of PDI, we used circular dichroism to examine the structural properties of PDI subjected to thermal denaturation. PDI exhibited remarkable conformational resilience that is linked to its redox status. In the reduced state, PDI exhibited a 54 °C unfolding transition temperature (Tm) and regained 85% of its native structure after nearly complete thermal denaturation. Oxidized PDI had a lower Tm of 48-50 °C and regained 70% of its native conformation after 75% denaturation. Both reduced PDI and oxidized PDI were functional after refolding from these denatured states. Additional studies documented increased stability of a PDI construct lacking the a' domain and decreased thermal stability of a construct lacking the a domain. Furthermore, oxidation of the a domain limited the ability of PDI to refold. The stability and conformational resilience of PDI are thus linked to both redox-dependent and domain-specific effects. These findings document previously unrecognized properties of PDI and provide insight into the physical foundation of its biological function.
Collapse
Affiliation(s)
- Jessica Guyette
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816 USA
| | - Baggio Evangelista
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816 USA
| | - Suren A. Tatulian
- Department of Physics, University of Central Florida, Orlando, FL 32816 USA
| | - Ken Teter
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816 USA
| |
Collapse
|
57
|
Hypoxia Modulates Effects of Fatty Acids on NES2Y Human Pancreatic β-cells. Int J Mol Sci 2019; 20:ijms20143441. [PMID: 31336948 PMCID: PMC6678120 DOI: 10.3390/ijms20143441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/11/2019] [Indexed: 12/27/2022] Open
Abstract
Saturated fatty acids (FAs) induce apoptosis in the human pancreatic NES2Y β-cell line while unsaturated FAs have nearly no detrimental effect. Moreover, unsaturated FAs are capable of inhibiting the pro-apoptotic effect of saturated FAs. Hypoxia is also known to have deleterious effects on β-cells function and viability. In the present study, we have tested the modulatory effect of hypoxia on the effect of FAs on the growth and viability of the human pancreatic NES2Y β-cells. This study represents the first study testing hypoxia effect on effects of FAs in pancreatic β-cells as well as in other cell types. We showed that hypoxia increased the pro-apoptotic effect of saturated stearic acid (SA). Endoplasmic reticulum stress signaling seemed to be involved while redistribution of FA transporters fatty acid translocase/cluster of differentiation 36 (FAT/CD36) and fatty acid-binding protein (FABP) do not seem to be involved in this effect. Hypoxia also strongly decreased the protective effect of unsaturated oleic acid (OA) against the pro-apoptotic effect of SA. Thus, in the presence of hypoxia, OA was unable to save SA-treated β-cells from apoptosis induction. Hypoxia itself had only a weak detrimental effect on NES2Y cells. Our data suggest that hypoxia could represent an important factor in pancreatic β-cell death induced and regulated by FAs and thus in the development of type 2 diabetes mellitus.
Collapse
|
58
|
Passam FJ, Chiu J. Allosteric disulphide bonds as reversible mechano-sensitive switches that control protein functions in the vasculature. Biophys Rev 2019; 11:419-430. [PMID: 31090016 DOI: 10.1007/s12551-019-00543-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/29/2019] [Indexed: 01/02/2023] Open
Abstract
Disulphide bonds are covalent linkages of two cysteine residues (R-S-S-R') in proteins. Unlike peptide bonds, disulphide bonds are reversible in nature allowing cleaved bonds to reform. Disulphide bonds are important structural elements that stabilise protein conformation. They can be of catalytic function found in enzymes that facilitate redox reactions in the cleavage/formation of disulphide bonds in their substrates. Emerging evidence also indicates that disulphide bonds can be of regulatory function which alter protein activity when they are cleaved or formed. This class of regulatory disulphide bonds is known as allosteric disulphide bonds. Allosteric disulphide bonds are mechano-sensitive, and stretching or twisting the sulphur-sulphur bond by mechanical force can make it easier or harder to be cleaved. This makes allosteric disulphide bonds an ideal type of mechano-sensitive switches for regulating protein functions in the vasculature where cells are continuously subjected to fluid shear force. This review will discuss the chemistry and biophysical properties of allosteric disulphide bonds and how they emerge to be mechano-sensitive switches in regulating platelet function and clot formation.
Collapse
Affiliation(s)
- Freda J Passam
- Heart Research Institute and Charles Perkins centre, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Joyce Chiu
- The Centenary Institute, NHMRC Clinical Trial Centre, Sydney Medical School, University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
59
|
Yogalakshmi B, Sathiya Priya C, Anuradha CV. Grape seed proanthocyanidins and metformin combination attenuate hepatic endoplasmic reticulum stress in rats subjected to nutrition excess. Arch Physiol Biochem 2019; 125:174-183. [PMID: 29482356 DOI: 10.1080/13813455.2018.1444058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT Endoplasmic reticulum (ER) stress in the liver is a pathological outcome of nutrient excess and is suggested to be one of the hits for progressive liver injury. OBJECTIVE This study investigated whether grape seed proanthocyanidins (GSP) and metformin (MET) alone or in combination can relieve hepatic ER stress induced in rats subjected to calorie excess. MATERIAL AND METHODS Male albino Wistar rats were given high calorie diet (HCD) for 45 days, while GSP (100 mg/kg body weight) and MET (50 mg/kg body weight) were administered either alone or in combination for last 15 days. RESULTS GSP, MET or both had reduced the levels of ER stress markers and chaperons, and suppressed the activation of lipogenic and inflammatory mediators in rat liver. DISCUSSION Though GSP and MET had reduced ER stress and inflammation individually, combination treatment with GSP + MET was more effective. CONCLUSION We suggest intervention with GSP and MET intake has to be considered for the management of liver disorders.
Collapse
Affiliation(s)
- Baskaran Yogalakshmi
- a Department of Biochemistry and Biotechnology , Annamalai University , Annamalai Nagar , India
| | | | | |
Collapse
|
60
|
Abstract
PURPOSE OF REVIEW The present review provides an overview of recent findings on new members of the protein disulfide isomerase (PDI) family required for thrombosis. RECENT FINDINGS Twenty years ago PDI was shown to mediate platelet aggregation, and 10 years ago PDI was shown to support thrombosis in vivo. Subsequently, other members of this endoplasmic reticulum family of enzymes, ERp57 and ERp5, were demonstrated to support thrombosis. A fourth member, ERp72, was recently shown to be required for platelet accumulation and fibrin deposition in vivo. None of these enzymes can individually support these processes. Moreover, aggregation of platelets deficient in a specific PDI is only recovered by the PDI that is missing. This implies that each PDI has a distinct role in activation of the αIIbβ3 fibrinogen receptor and platelet aggregation. Free thiols can be labeled in both subunits of αIIbβ3, suggesting cysteine-based reactions are involved in relaying conformational changes from the cytoplasmic tails to the integrin headpiece of this integrin. SUMMARY Multiple members of the PDI family support platelet function, and hemostasis and thrombosis with distinct roles in these processes. The individual cysteine targets of each enzyme and how these enzymes are integrated into a network that supports hemostasis and thrombosis remain to be elucidated.
Collapse
|
61
|
Matsusaki M, Kanemura S, Kinoshita M, Lee YH, Inaba K, Okumura M. The Protein Disulfide Isomerase Family: from proteostasis to pathogenesis. Biochim Biophys Acta Gen Subj 2019; 1864:129338. [PMID: 30986509 DOI: 10.1016/j.bbagen.2019.04.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/08/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
In mammalian cells, nearly one-third of proteins are inserted into the endoplasmic reticulum (ER), where they undergo oxidative folding and chaperoning assisted by approximately 20 members of the protein disulfide isomerase family (PDIs). PDIs consist of multiple thioredoxin-like domains and recognize a wide variety of proteins via highly conserved interdomain flexibility. Although PDIs have been studied intensely for almost 50 years, exactly how they maintain protein homeostasis in the ER remains unknown, and is important not only for fundamental biological understanding but also for protein misfolding- and aggregation-related pathophysiology. Herein, we review recent advances in structural biology and biophysical approaches that explore the underlying mechanism by which PDIs fulfil their distinct functions to promote productive protein folding and scavenge misfolded proteins in the ER, the primary factory for efficient production of the secretome.
Collapse
Affiliation(s)
- Motonori Matsusaki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shingo Kanemura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan; School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda, Hyogo 669-1337, Japan
| | - Misaki Kinoshita
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Young-Ho Lee
- Protein Structure Group, Korea Basic Science Institute, Ochang, Chungbuk 28199, South Korea; Bio-Analytical Science, University of Science and Technology, Daejeon 34113, South Korea
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai, Miyagi 980-8577, Japan.
| | - Masaki Okumura
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
62
|
Sutoh S, Uemura Y, Yamaguchi Y, Kiyotou A, Sugihara R, Nagayasu M, Kurokawa M, Ito K, Tsunekawa N, Nemoto M, Inagaki K, Tamura T. Redox-tuning of oxidizing disulfide oxidoreductase generates a potent disulfide isomerase. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:194-201. [DOI: 10.1016/j.bbapap.2018.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/27/2018] [Accepted: 12/14/2018] [Indexed: 11/16/2022]
|
63
|
Oliveira PVSD, Garcia-Rosa S, Sachetto ATA, Moretti AIS, Debbas V, De Bessa TC, Silva NT, Pereira ADC, Martins-de-Souza D, Santoro ML, Laurindo FRM. Protein disulfide isomerase plasma levels in healthy humans reveal proteomic signatures involved in contrasting endothelial phenotypes. Redox Biol 2019; 22:101142. [PMID: 30870787 PMCID: PMC6430080 DOI: 10.1016/j.redox.2019.101142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/22/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
Redox-related plasma proteins are candidate reporters of protein signatures associated with endothelial structure/function. Thiol-proteins from protein disulfide isomerase (PDI) family are unexplored in this context. Here, we investigate the occurrence and physiological significance of a circulating pool of PDI in healthy humans. We validated an assay for detecting PDI in plasma of healthy individuals. Our results indicate high inter-individual (median = 330 pg/mL) but low intra-individual variability over time and repeated measurements. Remarkably, plasma PDI levels could discriminate between distinct plasma proteome signatures, with PDI-rich (>median) plasma differentially expressing proteins related to cell differentiation, protein processing, housekeeping functions and others, while PDI-poor plasma differentially displayed proteins associated with coagulation, inflammatory responses and immunoactivation. Platelet function was similar among individuals with PDI-rich vs. PDI-poor plasma. Remarkably, such protein signatures closely correlated with endothelial function and phenotype, since cultured endothelial cells incubated with PDI-poor or PDI-rich plasma recapitulated gene expression and secretome patterns in line with their corresponding plasma signatures. Furthermore, such signatures translated into functional responses, with PDI-poor plasma promoting impairment of endothelial adhesion to fibronectin and a disturbed pattern of wound-associated migration and recovery area. Patients with cardiovascular events had lower PDI levels vs. healthy individuals. This is the first study describing PDI levels as reporters of specific plasma proteome signatures directly promoting contrasting endothelial phenotypes and functional responses.
Collapse
Affiliation(s)
- Percíllia Victória Santos de Oliveira
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Sheila Garcia-Rosa
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Ana Iochabel Soares Moretti
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Victor Debbas
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Tiphany Coralie De Bessa
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Nathalia Tenguan Silva
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
64
|
De Bessa TC, Pagano A, Moretti AIS, Oliveira PVS, Mendonça SA, Kovacic H, Laurindo FRM. Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas. Cell Death Dis 2019; 10:143. [PMID: 30760703 PMCID: PMC6374413 DOI: 10.1038/s41419-019-1402-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Protein disulfide isomerases including PDIA1 are implicated in cancer progression, but underlying mechanisms are unclear. PDIA1 is known to support vascular Nox1 NADPH oxidase expression/activation. Since deregulated reactive oxygen species (ROS) production underlies tumor growth, we proposed that PDIA1 is an upstream regulator of tumor-associated ROS. We focused on colorectal cancer (CRC) with distinct KRas activation levels. Analysis of RNAseq databanks and direct validation indicated enhanced PDIA1 expression in CRC with constitutive high (HCT116) vs. moderate (HKE3) and basal (Caco2) Ras activity. PDIA1 supported Nox1-dependent superoxide production in CRC; however, we first reported a dual effect correlated with Ras-level activity: in Caco2 and HKE3 cells, loss-of-function experiments indicate that PDIA1 sustains Nox1-dependent superoxide production, while in HCT116 cells PDIA1 restricted superoxide production, a behavior associated with increased Rac1 expression/activity. Transfection of Rac1G12V active mutant into HKE3 cells induced PDIA1 to become restrictive of Nox1-dependent superoxide, while in HCT116 cells treated with Rac1 inhibitor, PDIA1 became supportive of superoxide. PDIA1 silencing promoted diminished cell proliferation and migration in HKE3, not detectable in HCT116 cells. Screening of cell signaling routes affected by PDIA1 silencing highlighted GSK3β and Stat3. Also, E-cadherin expression after PDIA1 silencing was decreased in HCT116, consistent with PDIA1 support of epithelial-mesenchymal transition. Thus, Ras overactivation switches the pattern of PDIA1-dependent Rac1/Nox1 regulation, so that Ras-induced PDIA1 bypass can directly activate Rac1. PDIA1 may be a crucial regulator of redox-dependent adaptive processes related to cancer progression.
Collapse
Affiliation(s)
- Tiphany Coralie De Bessa
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Alessandra Pagano
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France
| | - Ana Iochabel Soares Moretti
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Percillia Victoria Santos Oliveira
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Samir Andrade Mendonça
- Centro de Investigação Translacional em Oncologia do Instituto do Câncer do Estado de São Paulo (Icesp), Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Herve Kovacic
- Aix Marseille Univ, CNRS, UMR 7051, INP, Inst Neurophysiopathol, Faculté de Pharmacie, 27, Boulevard Jean Moulin - 13385 Marseille CEDEX 5-France, Marseille, France.
| | - Francisco Rafael Martins Laurindo
- LIM 64, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
65
|
Meyer AJ, Riemer J, Rouhier N. Oxidative protein folding: state-of-the-art and current avenues of research in plants. THE NEW PHYTOLOGIST 2019; 221:1230-1246. [PMID: 30230547 DOI: 10.1111/nph.15436] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 08/01/2018] [Indexed: 06/08/2023]
Abstract
Contents Summary 1230 I. Introduction 1230 II. Formation and isomerization of disulfides in the ER and the Golgi apparatus 1231 III. The disulfide relay in the mitochondrial intermembrane space: why are plants different? 1236 IV. Disulfide bond formation on luminal proteins in thylakoids 1240 V. Conclusion 1242 Acknowledgements 1242 References 1242 SUMMARY: Disulfide bonds are post-translational modifications crucial for the structure and function of thousands of proteins. Their formation and isomerization, referred to as oxidative folding, require specific protein machineries found in oxidizing subcellular compartments, namely the endoplasmic reticulum and the associated endomembrane system, the intermembrane space of mitochondria and the thylakoid lumen of chloroplasts. At least one protein component is required for transferring electrons from substrate proteins to an acceptor that is usually molecular oxygen. For oxidation reactions, incoming reduced substrates are oxidized by thiol-oxidoreductase proteins (or domains in case of chimeric proteins), which are usually themselves oxidized by a single thiol oxidase, the enzyme generating disulfide bonds de novo. By contrast, the description of the molecular actors and pathways involved in proofreading and isomerization of misfolded proteins, which require a tightly controlled redox balance, lags behind. Herein we provide a general overview of the knowledge acquired on the systems responsible for oxidative protein folding in photosynthetic organisms, highlighting their particularities compared to other eukaryotes. Current research challenges are discussed including the importance and specificity of these oxidation systems in the context of the existence of reducing systems in the same compartments.
Collapse
Affiliation(s)
- Andreas J Meyer
- INRES-Chemical Signalling, University of Bonn, 53113, Bonn, Germany
| | - Jan Riemer
- Institute of Biochemistry, University of Cologne, 50674, Cologne, Germany
| | | |
Collapse
|
66
|
Javitt G, Grossman‐Haham I, Alon A, Resnick E, Mutsafi Y, Ilani T, Fass D. cis-Proline mutants of quiescin sulfhydryl oxidase 1 with altered redox properties undermine extracellular matrix integrity and cell adhesion in fibroblast cultures. Protein Sci 2019; 28:228-238. [PMID: 30367560 PMCID: PMC6295897 DOI: 10.1002/pro.3537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 11/13/2022]
Abstract
The thioredoxin superfamily has expanded and diverged extensively throughout evolution such that distant members no longer show appreciable sequence homology. Nevertheless, redox-active thioredoxin-fold proteins functioning in diverse physiological contexts often share canonical amino acids near the active-site (di-)cysteine motif. Quiescin sulfhydryl oxidase 1 (QSOX1), a catalyst of disulfide bond formation secreted by fibroblasts, is a multi-domain thioredoxin superfamily enzyme with certain similarities to the protein disulfide isomerase (PDI) enzymes. Among other potential functions, QSOX1 supports extracellular matrix assembly in fibroblast cultures. We introduced mutations at a cis-proline in QSOX1 that is conserved across the thioredoxin superfamily and was previously observed to modulate redox interactions of the bacterial enzyme DsbA. The resulting QSOX1 variants showed a striking detrimental effect when added exogenously to fibroblasts: they severely disrupted the extracellular matrix and cell adhesion, even in the presence of naturally secreted, wild-type QSOX1. The specificity of this phenomenon for particular QSOX1 mutants inspired an investigation of the effects of mutation on catalytic and redox properties. For a series of QSOX1 mutants, the detrimental effect correlated with the redox potential of the first redox-active site, and an X-ray crystal structure of one of the mutants revealed the reorganization of the cis-proline loop caused by the mutations. Due to the conservation of the mutated residues across the PDI family and beyond, insights obtained in this study may be broadly applicable to a variety of physiologically important redox-active enzymes. IMPACT STATEMENT: We show that mutation of a conserved cis-proline amino acid, analogous to a mutation used to trap substrates of a bacterial disulfide catalyst, has a dramatic effect on the physiological function of the mammalian disulfide catalyst QSOX1. As the active-site region of QSOX1 is shared with the large family of protein disulfide isomerases in humans, the effects of such mutations on redox properties, enzymatic activity, and biological targeting may be relevant across the family.
Collapse
Affiliation(s)
- Gabriel Javitt
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Iris Grossman‐Haham
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Assaf Alon
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Efrat Resnick
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Yael Mutsafi
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Tal Ilani
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Deborah Fass
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| |
Collapse
|
67
|
Stopa JD, Zwicker JI. The intersection of protein disulfide isomerase and cancer associated thrombosis. Thromb Res 2018; 164 Suppl 1:S130-S135. [PMID: 29703471 DOI: 10.1016/j.thromres.2018.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 01/08/2023]
Abstract
The mechanisms underlying the hypercoagulability of cancer are complex and include the upregulation coagulation factors or procoagulant proteins, shedding of microparticles, and direct activation of vascular cells. Protein disulfide isomerase (PDI) is a thiol isomerase secreted from activated platelets and endothelial cells and plays a critical role in both platelet aggregation and fibrin generation. A number of potential intravascular targets of PDI have been identified including cell surface receptors (e.g. β-integrins and glycoprotein Ib), receptor ligands (e.g. fibrinogen and von Willebrand factor), serine proteases (e.g. cathepsin G and kallekrein-14), and coagulation factors (e.g. factor XI and factor V). Recent clinical studies demonstrated that a small molecule inhibitor of PDI, isoquercetin, decreases platelet-dependent thrombin generation and PDI activity in plasma following oral administration. This review explores the mechanistic overlap between the molecular drivers of cancer associated thrombosis and the potential roles PDI plays in mediating thrombosis. These molecular insights provide rationale for clinical trials targeting PDI to prevent thrombosis in cancer patients.
Collapse
Affiliation(s)
- Jack D Stopa
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jeffrey I Zwicker
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
68
|
Protein disulfide isomerase does not act as an unfoldase in the disassembly of cholera toxin. Biosci Rep 2018; 38:BSR20181320. [PMID: 30135140 PMCID: PMC6127674 DOI: 10.1042/bsr20181320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/18/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022] Open
Abstract
Cholera toxin (CT) is composed of a disulfide-linked A1/A2 heterodimer and a ring-like, cell-binding B homopentamer. The catalytic A1 subunit must dissociate from CTA2/CTB5 to manifest its cellular activity. Reduction of the A1/A2 disulfide bond is required for holotoxin disassembly, but reduced CTA1 does not spontaneously separate from CTA2/CTB5: protein disulfide isomerase (PDI) is responsible for displacing CTA1 from its non-covalent assembly in the CT holotoxin. Contact with PDI shifts CTA1 from a protease-resistant conformation to a protease-sensitive conformation, which is thought to represent the PDI-mediated unfolding of CTA1. Based solely on this finding, PDI is widely viewed as an ‘unfoldase’ that triggers toxin disassembly by unfolding the holotoxin-associated A1 subunit. In contrast with this unfoldase model of PDI function, we report the ability of PDI to render CTA1 protease-sensitive is unrelated to its role in toxin disassembly. Multiple conditions that promoted PDI-induced protease sensitivity in CTA1 did not support PDI-mediated disassembly of the CT holotoxin. Moreover, preventing the PDI-induced shift in CTA1 protease sensitivity did not affect PDI-mediated disassembly of the CT holotoxin. Denatured PDI could still convert CTA1 into a protease-sensitive state, and equal or excess molar fractions of PDI were required for both efficient conversion of CTA1 into a protease-sensitive state and efficient disassembly of the CT holotoxin. These observations indicate the ‘unfoldase’ property of PDI does not play a functional role in CT disassembly and does not represent an enzymatic activity.
Collapse
|
69
|
The reductase TMX1 contributes to ERAD by preferentially acting on membrane-associated folding-defective polypeptides. Biochem Biophys Res Commun 2018; 503:938-943. [DOI: 10.1016/j.bbrc.2018.06.099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/19/2018] [Indexed: 11/22/2022]
|
70
|
Lamriben L, Oster ME, Tamura T, Tian W, Yang Z, Clausen H, Hebert DN. EDEM1's mannosidase-like domain binds ERAD client proteins in a redox-sensitive manner and possesses catalytic activity. J Biol Chem 2018; 293:13932-13945. [PMID: 30021839 DOI: 10.1074/jbc.ra118.004183] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/26/2018] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum (ER) degradation-enhancing α-mannosidase-like 1 protein (EDEM1) is a protein quality control factor that was initially proposed to recognize N-linked glycans on misfolded proteins through its mannosidase-like domain (MLD). However, recent studies have demonstrated that EDEM1 binds to some misfolded proteins in a glycan-independent manner, suggesting a more complex binding landscape for EDEM1. In this study, we have identified a thiol-dependent substrate interaction between EDEM1 and the α1-antitrypsin ER-associated protein degradation (ERAD) clients Z and NHK, specifically through the single Cys residue on Z/NHK (Cys256), required for binding under stringent detergent conditions. In addition to the thiol-dependent interaction, the presence of weaker protein-protein interactions was confirmed, suggestive of bipartite client-binding properties. About four reactive thiols on EDEM1 were identified and were not directly responsible for the observed redox-sensitive binding by EDEM1. Moreover, a protein construct comprising the EDEM1 MLD had thiol-dependent binding properties along with its active glycan-trimming activities. Lastly, we identified an additional intrinsically disordered region (IDR) located at the C terminus of EDEM1 in addition to its previously identified N-terminal IDR. We also determined that both IDRs are required for binding to the ERAD component ERdj5 as an interaction with ERdj5 was not observed with the MLD alone. Together, our findings indicate that EDEM1 employs different binding modalities to interact with ERAD clients and ER quality control (ERQC) machinery partners and that some of these properties are shared with its homologues EDEM2 and EDEM3.
Collapse
Affiliation(s)
- Lydia Lamriben
- From the Department of Biochemistry and Molecular Biology and.,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Michela E Oster
- From the Department of Biochemistry and Molecular Biology and
| | - Taku Tamura
- From the Department of Biochemistry and Molecular Biology and
| | - Weihua Tian
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Daniel N Hebert
- From the Department of Biochemistry and Molecular Biology and .,Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003 and
| |
Collapse
|
71
|
Fradin T, Bechor E, Berdichevsky Y, Dahan I, Pick E. Binding of p67phoxto Nox2 is stabilized by disulfide bonds between cysteines in the369Cys-Gly-Cys371triad in Nox2 and in p67phox. J Leukoc Biol 2018; 104:1023-1039. [DOI: 10.1002/jlb.4a0418-173r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/09/2018] [Accepted: 06/23/2018] [Indexed: 01/05/2023] Open
Affiliation(s)
- Tanya Fradin
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Edna Bechor
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Yevgeny Berdichevsky
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Iris Dahan
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| | - Edgar Pick
- The Julius Friedrich Cohnheim Laboratory of Phagocyte Research, Department of Clinical Microbiology and Immunology; Sackler School of Medicine, Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
72
|
Yu S, Ito S, Wada I, Hosokawa N. ER-resident protein 46 (ERp46) triggers the mannose-trimming activity of ER degradation-enhancing α-mannosidase-like protein 3 (EDEM3). J Biol Chem 2018; 293:10663-10674. [PMID: 29784879 DOI: 10.1074/jbc.ra118.003129] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/16/2018] [Indexed: 11/06/2022] Open
Abstract
Protein folding in the cell is regulated by several quality-control mechanisms. Correct folding of glycoproteins in the endoplasmic reticulum (ER) is tightly monitored by the recognition of glycan signals by lectins in the ER-associated degradation (ERAD) pathway. In mammals, mannose trimming from N-glycans is crucial for disposal of misfolded glycoproteins. The mannosidases responsible for this process are ER mannosidase I and ER degradation-enhancing α-mannosidase-like proteins (EDEMs). However, the molecular mechanism of mannose removal by EDEMs remains unclear, partly owing to the difficulty of reconstituting mannosidase activity in vitro Here, our analysis of EDEM3-mediated mannose-trimming activity on a misfolded glycoprotein revealed that ERp46, an ER-resident oxidoreductase, associates stably with EDEM3. This interaction, which depended on the redox activity of ERp46, involved formation of a disulfide bond between the cysteine residues of the ERp46 redox-active sites and the EDEM3 α-mannosidase domain. In a defined in vitro system consisting of recombinant proteins purified from HEK293 cells, the mannose-trimming activity of EDEM3 toward the model misfolded substrate, the glycoprotein T-cell receptor α locus (TCRα), was reconstituted only when ERp46 had established a covalent interaction with EDEM3. On the basis of these findings, we propose that disposal of misfolded glycoproteins through mannose trimming is tightly connected to redox-mediated regulation in the ER.
Collapse
Affiliation(s)
- Shangyu Yu
- From the Laboratory of Molecular and Cellular Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507
| | - Shinji Ito
- the Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, and
| | - Ikuo Wada
- the Department of Cell Sciences, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Nobuko Hosokawa
- From the Laboratory of Molecular and Cellular Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507,
| |
Collapse
|
73
|
Chen F, Zhao Z, Zhou J, Lu Y, Essex DW, Wu Y. Protein disulfide isomerase enhances tissue factor-dependent thrombin generation. Biochem Biophys Res Commun 2018; 501:172-177. [PMID: 29709484 DOI: 10.1016/j.bbrc.2018.04.207] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/26/2018] [Indexed: 01/01/2023]
Abstract
Protein disulfide isomerase (PDI) plays an important role in fibrin generation in vivo, but the underlying mechanism remains largely unknown. In this study, using thrombin generation assay (TGA), we investigated whether PDI contributes to tissue factor (TF)-mediated thrombin generation. Human peripheral blood mononuclear cells (PBMCs) were treated with 100 ng/ml lipopolysaccharide (LPS), the expression of TF on cell surface was analyzed by flow cytometry. After incubation with an inhibitory anti-TF antibody, recombinant PDI protein or a PDI inhibitor PACMA31, LPS-stimulated human PBMCs were incubated with human plasma, and thrombin generation was assessed by Ceveron Alpha TGA and a fluorescent thrombin substrate. Bone marrow mononuclear cells isolated from PDI-knockout and wild-type mice were stimulated by LPS, followed by measurement of thrombin generation. LPS stimulation increased expression of TF on PBMCs, and thrombin generation. Inhibitory anti-TF antibody almost completely suppressed thrombin generation of LPS-stimulated PBMCs, suggesting that thrombin generation was TF-dependent. Recombinant PDI protein increased thrombin generation, while PACMA31 attenuated thrombin generation. Compared with control cells, PDI-deficient marrow mononuclear cells had less capacity in thrombin generation. Taken together, these data suggest that PDI enhances TF-dependent thrombin generation.
Collapse
Affiliation(s)
- Fengwu Chen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhenzhen Zhao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Junsong Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yi Lu
- Wuhan Thalys Medical Technology Inc, Wuhan, China
| | - David W Essex
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA19140, USA
| | - Yi Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA19140, USA.
| |
Collapse
|
74
|
Abstract
The protein disulfide isomerase (PDI) family is a group of multifunctional endoplasmic reticulum (ER) enzymes that mediate the formation of disulfide bonds, catalyze the cysteine-based redox reactions and assist the quality control of client proteins. Recent structural and functional studies have demonstrated that PDI members not only play an essential role in the proteostasis in the ER but also exert diverse effects in numerous human disorders including cancer and neurodege-nerative diseases. Increasing evidence suggests that PDI is actively involved in the proliferation, survival, and metastasis of several types of cancer cells. Although the molecular mechanism by which PDI contributes to tumorigenesis and metastasis remains to be understood, PDI is now emerging as a new therapeutic target for cancer treatment. In fact, several attempts have been made to develop PDI inhibitors as anti-cancer drugs. In this review, we discuss the properties and diverse functions of human PDI proteins and focus on recent findings regarding their roles in the state of diseases including cancer and neurodegeneration.
Collapse
Affiliation(s)
- Eunyoug Lee
- Department of Bio and Environmental Technology, Seoul Women's University, Seoul 01797, Korea
| | - Do Hee Lee
- Department of Bio and Environmental Technology, Seoul Women's University, Seoul 01797, Korea
| |
Collapse
|
75
|
Schaefer K, Webb NE, Pang M, Hernandez-Davies JE, Lee KP, Gonzalez P, Douglass MV, Lee B, Baum LG. Galectin-9 binds to O-glycans on protein disulfide isomerase. Glycobiology 2018; 27:878-887. [PMID: 28810662 DOI: 10.1093/glycob/cwx065] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
Changes in the T cell surface redox environment regulate critical cell functions, such as cell migration, viral entry and cytokine production. Cell surface protein disulfide isomerase (PDI) contributes to the regulation of T cell surface redox status. Cell surface PDI can be released into the extracellular milieu or can be internalized by T cells. We have found that galectin-9, a soluble lectin expressed by T cells, endothelial cells and dendritic cells, binds to and retains PDI on the cell surface. While endogenous galectin-9 is not required for basal cell surface PDI expression, exogenous galectin-9 mediated retention of cell surface PDI shifted the disulfide/thiol equilibrium on the T cell surface. O-glycans on PDI are required for galectin-9 binding, and PDI recognition appears to be specific for galectin-9, as galectin-1 and galectin-3 do not bind PDI. Galectin-9 is widely expressed by immune and endothelial cells in inflamed tissues, suggesting that T cells would be exposed to abundant galectin-9, in cis and in trans, in infectious or autoimmune conditions.
Collapse
Affiliation(s)
| | - Nicholas E Webb
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Mabel Pang
- Department of Pathology and Laboratory Medicine
| | | | | | | | | | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine, Mount Sinai, New York, USA
| | | |
Collapse
|
76
|
Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, Chauhan SC. Antibody-Drug Conjugates for Cancer Therapy: Chemistry to Clinical Implications. Pharmaceuticals (Basel) 2018; 11:ph11020032. [PMID: 29642542 PMCID: PMC6027311 DOI: 10.3390/ph11020032] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023] Open
Abstract
Chemotherapy is one of the major therapeutic options for cancer treatment. Chemotherapy is often associated with a low therapeutic window due to its poor specificity towards tumor cells/tissues. Antibody-drug conjugate (ADC) technology may provide a potentially new therapeutic solution for cancer treatment. ADC technology uses an antibody-mediated delivery of cytotoxic drugs to the tumors in a targeted manner, while sparing normal cells. Such a targeted approach can improve the tumor-to-normal tissue selectivity and specificity in chemotherapy. Considering its importance in cancer treatment, we aim to review recent efforts for the design and development of ADCs. ADCs are mainly composed of an antibody, a cytotoxic payload, and a linker, which can offer selectivity against tumors, anti-cancer activity, and stability in systemic circulation. Therefore, we have reviewed recent updates and principal considerations behind ADC designs, which are not only based on the identification of target antigen, cytotoxic drug, and linker, but also on the drug-linker chemistry and conjugation site at the antibody. Our review focuses on site-specific conjugation methods for producing homogenous ADCs with constant drug-antibody ratio (DAR) in order to tackle several drawbacks that exists in conventional conjugation methods.
Collapse
Affiliation(s)
- Nirnoy Dan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Vivek K Kashyap
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sheema Khan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
77
|
Grosche J, Meißner J, Eble JA. More than a syllable in fib-ROS-is: The role of ROS on the fibrotic extracellular matrix and on cellular contacts. Mol Aspects Med 2018; 63:30-46. [PMID: 29596842 DOI: 10.1016/j.mam.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023]
Abstract
Fibrosis is characterized by excess deposition of extracellular matrix (ECM). However, the ECM changes during fibrosis not only quantitatively but also qualitatively. Thus, the composition is altered as the expression of various ECM proteins changes. Moreover, also posttranslational modifications, secretion, deposition and crosslinkage as well as the proteolytic degradation of ECM components run differently during fibrosis. As several of these processes involve redox reactions and some of them are even redox-regulated, reactive oxygen species (ROS) influence fibrotic diseases. Redox regulation of the ECM has not been studied intensively, although evidences exist that the alteration of the ECM, including the redox-relevant processes of its formation and degradation, may be of key importance not only as a cause but also as a consequence of fibrotic diseases. Myofibroblasts, which have differentiated from fibroblasts during fibrosis, produce most of the ECM components and in return obtain important environmental cues of the ECM, including their redox-dependent fibrotic alterations. Thus, myofibroblast differentiation and fibrotic changes of the ECM are interdependent processes and linked with each other via cell-matrix contacts, which are mediated by integrins and other cell adhesion molecules. These cell-matrix contacts are also regulated by redox processes and by ROS. However, most of the redox-catalyzing enzymes are localized within cells. Little is known about redox-regulating enzymes, especially the ones that control the formation and cleavage of redox-sensitive disulfide bridges within the extracellular space. They are also important players in the redox-regulative crosstalk between ECM and cells during fibrosis.
Collapse
Affiliation(s)
- Julius Grosche
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Juliane Meißner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany.
| |
Collapse
|
78
|
Liang Y, Li S, Wang X, Zhang Y, Sun Y, Wang Y, Wang X, He B, Dai W, Zhang H, Wang X, Zhang Q. A comparative study of the antitumor efficacy of peptide-doxorubicin conjugates with different linkers. J Control Release 2018; 275:129-141. [DOI: 10.1016/j.jconrel.2018.01.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/06/2018] [Accepted: 01/30/2018] [Indexed: 12/12/2022]
|
79
|
Abstract
INTRODUCTION The protein disulfide isomerase (PDI) family of thiol isomerases are intracellular enzymes known to catalyze the oxidation, reduction and isomerization of disulfide bonds during protein synthesis in the endoplasmic reticulum. PDI and related members of the thiol isomerase family are known to localize extracellularly where they possess various functions. Among these, the role of PDI in the initiation of thrombus formation is best characterized. PDI is secreted within seconds from activated platelets and endothelial cells at the site of vascular injury and accumulates in the developing platelet-fibrin thrombus. Inhibition of PDI by antibodies or small molecule inhibitors blocks thrombus formation. Efforts are underway to identify extracellular substrates of PDI that participate in the network pathways linking thiol isomerases to thrombus formation. ERp57, ERp5 and ERp72 also play a role in initiation of thrombus formation but their specific extracellular substrates are unknown. Areas covered: The following review gives an overview of biochemistry of vascular thiol isomerases followed by a detailed description of their role in thrombosis and its clinical implications. Expert commentary: The thiol isomerase system, by controlling the initiation of thrombus formation, provides the regulatory switch by which the normal vasculature is protected under physiologic conditions from thrombi generation.
Collapse
Affiliation(s)
- Anish Sharda
- a Division of Hemostasis and Thrombosis , Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA USA
| | - Bruce Furie
- a Division of Hemostasis and Thrombosis , Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA USA
| |
Collapse
|
80
|
Cole KS, Grandjean JMD, Chen K, Witt CH, O'Day J, Shoulders MD, Wiseman RL, Weerapana E. Characterization of an A-Site Selective Protein Disulfide Isomerase A1 Inhibitor. Biochemistry 2018. [PMID: 29521097 DOI: 10.1021/acs.biochem.8b00178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Protein disulfide isomerase A1 (PDIA1) is an endoplasmic reticulum (ER)-localized thiol-disulfide oxidoreductase that is an important folding catalyst for secretory pathway proteins. PDIA1 contains two active-site domains (a and a'), each containing a Cys-Gly-His-Cys (CGHC) active-site motif. The two active-site domains share 37% sequence identity and function independently to perform disulfide-bond reduction, oxidation, and isomerization. Numerous inhibitors for PDIA1 have been reported, yet the selectivity of these inhibitors toward the a and a' sites is poorly characterized. Here, we identify a potent and selective PDIA1 inhibitor, KSC-34, with 30-fold selectivity for the a site over the a' site. KSC-34 displays time-dependent inhibition of PDIA1 reductase activity in vitro with a kinact/ KI of 9.66 × 103 M-1 s-1 and is selective for PDIA1 over other members of the PDI family, and other cellular cysteine-containing proteins. We provide the first cellular characterization of an a-site selective PDIA1 inhibitor and demonstrate that KSC-34 has minimal sustained effects on the cellular unfolded protein response, indicating that a-site inhibition does not induce global protein folding-associated ER stress. KSC-34 treatment significantly decreases the rate of secretion of a destabilized, amyloidogenic antibody light chain, thereby minimizing pathogenic amyloidogenic extracellular proteins that rely on high PDIA1 activity for proper folding and secretion. Given the poor understanding of the contribution of each PDIA1 active site to the (patho)physiological functions of PDIA1, site selective inhibitors like KSC-34 provide useful tools for delineating the pathological role and therapeutic potential of PDIA1.
Collapse
Affiliation(s)
- Kyle S Cole
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Julia M D Grandjean
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Kenny Chen
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Collin H Witt
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Johanna O'Day
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| | - Matthew D Shoulders
- Department of Chemistry , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - R Luke Wiseman
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Eranthie Weerapana
- Department of Chemistry , Boston College , Chestnut Hill , Massachusetts 02467 , United States
| |
Collapse
|
81
|
Kim Y, Chang KO. Protein disulfide isomerases as potential therapeutic targets for influenza A and B viruses. Virus Res 2018; 247:26-33. [PMID: 29382552 PMCID: PMC5831498 DOI: 10.1016/j.virusres.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 01/03/2023]
Abstract
Seasonal flu as well as potential pandemic flu outbreaks continuously underscores the importance of the preventive and therapeutic measures against influenza viruses. During screening of natural and synthetic small molecules against influenza A and B virus, we identified juniferdin as a highly effective inhibitor against both viruses in cells. Since juniferdin is known to inhibit protein disulfide isomerases (PDIs), multiple PDI inhibitors were tested against these viruses. Among PDI inhibitors, 16F16, PACMA31, isoquercetin, epigallocatechin-3-gallate or nitazoxanide significantly reduced the replication of influenza A and B viruses in MDCK and A549 cells. Furthermore, siRNAs specific to three PDI family members (PDI1, PDIA3 or PDIA4) also significantly reduced the replication of influenza A and B viruses in cells. These results suggest that PDIs may serve as excellent targets for the development of new anti-influenza drugs.
Collapse
Affiliation(s)
- Yunjeong Kim
- Department of Pathobiology and Preventive Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Kyeong-Ok Chang
- Department of Pathobiology and Preventive Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
82
|
Caba C, Ali Khan H, Auld J, Ushioda R, Araki K, Nagata K, Mutus B. Conserved Residues Lys 57 and Lys 401 of Protein Disulfide Isomerase Maintain an Active Site Conformation for Optimal Activity: Implications for Post-Translational Regulation. Front Mol Biosci 2018. [PMID: 29541639 PMCID: PMC5835755 DOI: 10.3389/fmolb.2018.00018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Despite its study since the 1960's, very little is known about the post-translational regulation of the multiple catalytic activities performed by protein disulfide isomerase (PDI), the primary protein folding catalyst of the cell. This work identifies a functional role for the highly conserved CxxC-flanking residues Lys57 and Lys401 of human PDI in vitro. Mutagenesis studies have revealed these residues as modulating the oxidoreductase activity of PDI in a pH-dependent manner. Non-conservative amino acid substitutions resulted in enzyme variants upwards of 7-fold less efficient. This attenuated activity was found to translate into a 2-fold reduction of the rate of electron shuttling between PDI and the intraluminal endoplasmic reticulum oxidase, ERO1α, suggesting a functional significance to oxidative protein folding. In light of this, the possibility of lysine acetylation at residues Lys57 and Lys401 was assessed by in vitro treatment using acetylsalicylic acid (aspirin). A total of 28 acetyllysine residues were identified, including acLys57 and acLys401. The kinetic behavior of the acetylated protein form nearly mimicked that obtained with a K57/401Q double substitution variant providing an indication that acetylation of the active site-flanking lysine residues can act to reversibly modulate PDI activity.
Collapse
Affiliation(s)
- Cody Caba
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Hyder Ali Khan
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Janeen Auld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Ryo Ushioda
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Kazutaka Araki
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Kazuhiro Nagata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Bulent Mutus
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
83
|
Doron L, Goloubinoff P, Shapira M. ZnJ2 Is a Member of a Large Chaperone Family in the Chloroplast of Photosynthetic Organisms that Features a DnaJ-Like Zn-Finger Domain. Front Mol Biosci 2018; 5:2. [PMID: 29497613 PMCID: PMC5818400 DOI: 10.3389/fmolb.2018.00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/08/2018] [Indexed: 11/13/2022] Open
Abstract
Photosynthesis is performed by large complexes, composed of subunits encoded by the nuclear and chloroplast genomes. Assembly is assisted by general and target-specific chaperones, but their mode of action is yet unclear. We formerly showed that ZnJ2 is an algal chaperone resembling BSD2 from land plants. In algae, it co-migrates with the rbcL transcript on chloroplast polysomes, suggesting it contributes to the de-novo synthesis of RbcL (Doron et al., 2014). ZnJ2 contains four CXXCXGXG motifs, comprising a canonical domain typical also of DnaJ-type I (DNAJA). It contributes to the binding of protein substrates to DnaK and promotes an independent oxidoreductase activity (Mattoo et al., 2014). To examine whether ZnJ2 has oxidoreductase activity, we used the RNaseA assay, which measures the oxidation-dependent reactivation of reduced-denatured RNaseA. Although ZnJ2 assisted the native refolding of reduced-denatured RNaseA, its activity was restricted to an oxidizing environment. Thus, ZnJ2 did not carry the exclusive responsibility for the formation of disulfide bridges, but contributed to the stabilization of its target polypeptides, until they reached their native state. A ZnJ2 cysteine deficient mutant maintained a similar holding chaperone activity as the wild-type and did not induce the formation of disulfide bonds. ZnJ2 is devoid of a J-domain. It thus does not belong to the J-domain co-chaperones that target protein substrates to DnaK. As expected, in vitro, its aggregation-prevention activity was not synergic to the ATP-fueled action of DnaK/DnaJ/GrpE in assisting the native refolding of denatured malate dehydrogenase, nor did it show an independent refolding activity. A phylogenetic analysis showed that ZnJ2 and BSD2 from land plants, are two different proteins belonging to a larger group containing a cysteine-rich domain, that also includes the DNAJAs. Members of this family are apparently involved in specific assembly of photosynthetic complexes in the chloroplast.
Collapse
Affiliation(s)
- Lior Doron
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Pierre Goloubinoff
- Department of Plant Molecular Biology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Michal Shapira
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
84
|
Berner N, Reutter KR, Wolf DH. Protein Quality Control of the Endoplasmic Reticulum and Ubiquitin-Proteasome-Triggered Degradation of Aberrant Proteins: Yeast Pioneers the Path. Annu Rev Biochem 2018; 87:751-782. [PMID: 29394096 DOI: 10.1146/annurev-biochem-062917-012749] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells must constantly monitor the integrity of their macromolecular constituents. Proteins are the most versatile class of macromolecules but are sensitive to structural alterations. Misfolded or otherwise aberrant protein structures lead to dysfunction and finally aggregation. Their presence is linked to aging and a plethora of severe human diseases. Thus, misfolded proteins have to be rapidly eliminated. Secretory proteins constitute more than one-third of the eukaryotic proteome. They are imported into the endoplasmic reticulum (ER), where they are folded and modified. A highly elaborated machinery controls their folding, recognizes aberrant folding states, and retrotranslocates permanently misfolded proteins from the ER back to the cytosol. In the cytosol, they are degraded by the highly selective ubiquitin-proteasome system. This process of protein quality control followed by proteasomal elimination of the misfolded protein is termed ER-associated degradation (ERAD), and it depends on an intricate interplay between the ER and the cytosol.
Collapse
Affiliation(s)
- Nicole Berner
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| | - Karl-Richard Reutter
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| | - Dieter H Wolf
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany; , ,
| |
Collapse
|
85
|
Gao H, Sun B, Fu H, Chi X, Wang F, Qi X, Hu J, Shao S. PDIA6 promotes the proliferation of HeLa cells through activating the Wnt/β-catenin signaling pathway. Oncotarget 2018; 7:53289-53298. [PMID: 27462866 PMCID: PMC5288186 DOI: 10.18632/oncotarget.10795] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase family 6 (PDIA6) belongs to the protein disulfide isomerase (PDI) family, which function as isomerases and molecular chaperones. PDIA6 has recently been shown to promote the proliferation and growth of various types of human cancer cells; however the underlying molecular mechanism remains elusive. Here, we report that PDIA6 enhances the proliferation of HeLa cells through activation of the Wnt/β-catenin signaling pathway. Ectopic overexpression of PDIA6 in HeLa cells led to increased cell proliferation accompanied with accelerated cell cycle progression. Further mechanistic investigation demonstrated that overexpression of PDIA6 resulted in decreased phosphorylation of β-catenin at Ser45 and Ser33/Ser37/Thr41, while increased β-catenin nuclear accumulation, and upregulation of Wnt/ β-catenin signaling target genes cyclinD1 and c-myc, which was abolished by ubiquitin-proteasome inhibitor MG132. These results demonstrated that PDIA6 overexpression promoted the proliferation of HeLa cells by suppressing the phosphorylation of β-catenin, thereby inhibiting the degradation of β-catenin through the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Huijun Gao
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Bing Sun
- Department of Thoracic Surgery, The First Hospital of Dalian Medical University, Dalian, China
| | - Hailu Fu
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Xinming Chi
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Faming Wang
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Xiaoyu Qi
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Jun Hu
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | - Shujuan Shao
- Key Laboratory of Proteomics, Dalian Medical University, Dalian, China
| |
Collapse
|
86
|
Zhang P, Wu J, Xiao F, Zhao D, Luan Y. Disulfide bond based polymeric drug carriers for cancer chemotherapy and relevant redox environments in mammals. Med Res Rev 2018; 38:1485-1510. [PMID: 29341223 DOI: 10.1002/med.21485] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/14/2017] [Accepted: 12/26/2017] [Indexed: 12/14/2022]
Abstract
Increasing numbers of disulfide linkage-employing polymeric drug carriers that utilize the reversible peculiarity of this unique covalent bond have been reported. The reduction-sensitive disulfide bond is usually employed as a linkage between hydrophilic and hydrophobic polymers, polymers and drugs, or as cross-linkers in polymeric drug carriers. These polymeric drug carriers are designed to exploit the significant redox potential difference between the reducing intracellular environments and relatively oxidizing extracellular spaces. In addition, these drug carriers can release a considerable amount of anticancer drug in response to the reducing environment when they reach tumor tissues, effectively improving antitumor efficacy. This review focuses on various disulfide linkage-employing polymeric drug carriers. Important redox thiol pools, including GSH/GSSG, Cys/CySS, and Trx1, as well as redox environments in mammals, will be introduced.
Collapse
Affiliation(s)
- Pei Zhang
- School of Pharmaceutical Science, Shandong University, Jinan, P. R. China
| | - Jilian Wu
- School of Pharmaceutical Science, Shandong University, Jinan, P. R. China
| | - Fengmei Xiao
- Binzhou Tuberculosis Prevention and Treatment Hospital, Binzhou, P. R. China
| | - Dujuan Zhao
- School of Pharmaceutical Science, Shandong University, Jinan, P. R. China
| | - Yuxia Luan
- School of Pharmaceutical Science, Shandong University, Jinan, P. R. China
| |
Collapse
|
87
|
Slow domain reconfiguration causes power-law kinetics in a two-state enzyme. Proc Natl Acad Sci U S A 2018; 115:513-518. [PMID: 29298911 DOI: 10.1073/pnas.1714401115] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein dynamics are typically captured well by rate equations that predict exponential decays for two-state reactions. Here, we describe a remarkable exception. The electron-transfer enzyme quiescin sulfhydryl oxidase (QSOX), a natural fusion of two functionally distinct domains, switches between open- and closed-domain arrangements with apparent power-law kinetics. Using single-molecule FRET experiments on time scales from nanoseconds to milliseconds, we show that the unusual open-close kinetics results from slow sampling of an ensemble of disordered domain orientations. While substrate accelerates the kinetics, thus suggesting a substrate-induced switch to an alternative free energy landscape of the enzyme, the power-law behavior is also preserved upon electron load. Our results show that the slow sampling of open conformers is caused by a variety of interdomain interactions that imply a rugged free energy landscape, thus providing a generic mechanism for dynamic disorder in multidomain enzymes.
Collapse
|
88
|
Characterization of the oxidative protein folding activity of a unique plant oxidoreductase, Arabidopsis protein disulfide isomerase-11. Biochem Biophys Res Commun 2018; 495:1041-1047. [DOI: 10.1016/j.bbrc.2017.11.111] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 11/19/2022]
|
89
|
Wang X, Xue G, Song M, Xu P, Chen D, Yuan C, Lin L, Flaumenhaft R, Li J, Huang M. Molecular basis of rutin inhibition of protein disulfide isomerase (PDI) by combinedin silicoand experimental methods. RSC Adv 2018; 8:18480-18491. [PMID: 35541126 PMCID: PMC9080521 DOI: 10.1039/c8ra02683a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/02/2018] [Indexed: 01/13/2023] Open
Abstract
Protein disulfide isomerase (PDI) is a founding member of the thiol isomerase family, and is recently found to play critical roles in thrombus formation. The development of effective PDI inhibitors is of great significance, and attracts strong interest. We previously showed that rutin bound directly to PDI and inhibited PDI activities, leading to the suppression of platelet aggregation and fibrin generation in a mouse model. A close analog of rutin, isoquercetin, is currently in advanced phase clinical trials. However, the molecular interaction between rutin and PDI is unknown and is difficult to study by X-ray crystallography due to the weak interaction. Here, we generated a molecular model of PDI:rutin complex by molecular docking and thorough molecular dynamics (MD) simulations. We then validated the complex model through a number of different experimental methods. We mutated the key residues predicted by the model and analyzed the mutants by an optimized isothermal titration calorimetry (ITC) method and a functional assay (insulin reduction assay). The results consistently showed that the PDI residues H354, L355 and E359 are important in the binding of rutin. These residues are next to the canonical major substrate binding site of the b′ domain, and were not conserved across the members of thiol isomerases, explaining the specificity of rutin for PDI among vascular thiol isomerases. Furthermore, the inhibitory activities of three rutin analogues were evaluated using an insulin reduction assay. The results supported that the second sugar ring at the side chain of rutin was not necessary for the binding to PDI. Together, this work provides the structural basis for the inhibitory mechanism of rutin to PDI, and offers a promising strategy for the design of new generation inhibitors with higher binding affinity to PDI for therapeutic applications. Rutin binds and inhibits PDI at b′x domain, H354 is one of the main binding sites.![]()
Collapse
Affiliation(s)
- Xu Wang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | - Guangpu Xue
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Meiru Song
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Peng Xu
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | - Dan Chen
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Cai Yuan
- College of Biological Science and Engineering
- Fuzhou University
- Fuzhou 350116
- China
| | - Lin Lin
- Beth Israel Deaconess Medical Center
- Harvard Medical School
- Boston
- USA
| | | | - Jinyu Li
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Mingdong Huang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| |
Collapse
|
90
|
Targeting the enhanced ER stress response in Marinesco-Sjögren syndrome. J Neurol Sci 2017; 385:49-56. [PMID: 29406913 DOI: 10.1016/j.jns.2017.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 11/11/2017] [Accepted: 12/08/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Marinesco-Sjögren syndrome (MSS) is an autosomal recessive infantile-onset disorder characterized by cataracts, cerebellar ataxia, and progressive myopathy caused by mutation of SIL1. In mice, a defect in SIL1 causes endoplasmic reticulum (ER) chaperone dysfunction, leading to unfolded protein accumulation and increased ER stress. However, ER stress and the unfolded protein response (UPR) have not been investigated in MSS patient-derived cells. METHODS Lymphoblastoid cell lines (LCLs) were established from four MSS patients. Spontaneous and tunicamycin-induced ER stress and the UPR were investigated in MSS-LCLs. Expression of UPR markers was analyzed by western blotting. ER stress-induced apoptosis was analyzed by flow cytometry. The cytoprotective effects of ER stress modulators were also examined. RESULTS MSS-LCLs exhibited increased spontaneous ER stress and were highly susceptible to ER stress-induced apoptosis. The inositol-requiring protein 1α (IRE1α)-X-box-binding protein 1 (XBP1) pathway was mainly upregulated in MSS-LCLs. Tauroursodeoxycholic acid (TUDCA) attenuated ER stress-induced apoptosis. CONCLUSION MSS patient-derived cells exhibit increased ER stress, an activated UPR, and susceptibility to ER stress-induced death. TUDCA reduces ER stress-induced death of MSS patient-derived cells. The potential of TUDCA as a therapeutic agent for MSS could be explored further in preclinical studies.
Collapse
|
91
|
Manuel AM, Walla MD, Faccenda A, Martin SL, Tanis RM, Piroli GG, Adam J, Kantor B, Mutus B, Townsend DM, Frizzell N. Succination of Protein Disulfide Isomerase Links Mitochondrial Stress and Endoplasmic Reticulum Stress in the Adipocyte During Diabetes. Antioxid Redox Signal 2017; 27:1281-1296. [PMID: 28376661 PMCID: PMC5655420 DOI: 10.1089/ars.2016.6853] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Protein succination by fumarate increases in the adipose tissue of diabetic mice and in adipocytes matured in high glucose as a result of glucotoxicity-driven mitochondrial stress. The endoplasmic reticulum (ER) oxidoreductase protein disulfide isomerase (PDI) is succinated in adipocytes that are matured in high glucose, and in this study we investigated whether succination would alter PDI oxidoreductase activity, directly linking mitochondrial stress and ER stress. RESULTS Protein succination and the ER stress marker C/EBP homologous protein (CHOP) were diminished after pharmaceutical targeting of mitochondrial stress with the chemical uncoupler niclosamide in adipocytes matured in high-glucose concentrations. PDI was succinated by fumarate on both CXXC-containing active sites, contributing to reduced enzymatic activity. Succinated PDI decreased reductase activity in adipocytes matured in high glucose, and in db/db epididymal adipose tissue, in association with increased levels of CHOP. PDI succination was increased in fumarase knockdown adipocytes, leading to reduced PDI oxidoreductase activity, increased CHOP levels, and pro-inflammatory cytokine secretion, confirming the specific role of elevated fumarate levels in contributing to ER stress. In addition, PDI succination and ER stress were decreased, and PDI reductase activity was restored when exposure to chronic high glucose was limited, highlighting the importance of calorie restriction in the improvement of adipocyte metabolic function. INNOVATION These experiments identify PDI succination as a novel biochemical mechanism linking altered mitochondrial metabolism to ER stress in the adipocyte during diabetes. CONCLUSION The current study demonstrates that early biochemical changes in mitochondrial metabolism have important implications for the development of adipocyte stress. Antioxid. Redox Signal. 27, 1281-1296.
Collapse
Affiliation(s)
- Allison M Manuel
- 1 Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Michael D Walla
- 2 Mass Spectrometry Center, Department of Chemistry and Biochemistry, University of South Carolina , Columbia, South Carolina
| | - Adam Faccenda
- 3 Department of Chemistry and Biochemistry, University of Windsor , Windsor, Canada
| | - Stephanie L Martin
- 1 Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Ross M Tanis
- 1 Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Gerardo G Piroli
- 1 Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Julie Adam
- 4 Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford , Oxford, United Kingdom
| | - Boris Kantor
- 5 Viral Vector Core, Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Bulent Mutus
- 3 Department of Chemistry and Biochemistry, University of Windsor , Windsor, Canada
| | - Danyelle M Townsend
- 6 Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Norma Frizzell
- 1 Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina , Columbia, South Carolina
| |
Collapse
|
92
|
Delaunay-Moisan A, Ponsero A, Toledano MB. Reexamining the Function of Glutathione in Oxidative Protein Folding and Secretion. Antioxid Redox Signal 2017; 27:1178-1199. [PMID: 28791880 DOI: 10.1089/ars.2017.7148] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Disturbance of glutathione (GSH) metabolism is a hallmark of numerous diseases, yet GSH functions are poorly understood. One key to this question is to consider its functional compartmentation. GSH is present in the endoplasmic reticulum (ER), where it competes with substrates for oxidation by the oxidative folding machinery, composed in eukaryotes of the thiol oxidase Ero1 and proteins from the disulfide isomerase family (protein disulfide isomerase). Yet, whether GSH is required for proper ER oxidative protein folding is a highly debated question. Recent Advances: Oxidative protein folding has been thoroughly dissected over the past decades, and its actors and their mode of action elucidated. Genetically encoded GSH probes have recently provided an access to subcellular redox metabolism, including the ER. CRITICAL ISSUES Of the few often-contradictory models of the role of GSH in the ER, the most popular suggest it serves as reducing power. Yet, as a reductant, GSH also activates Ero1, which questions how GSH can nevertheless support protein reduction. Hence, whether GSH operates in the ER as a reductant, an oxidant, or just as a "blank" compound mirroring ER/periplasm redox activity is a highly debated question, which is further stimulated by the puzzling occurrence of GSH in the Escherichia coli periplasmic "secretory" compartment, aside from the Dsb thiol-reducing and oxidase pathways. FUTURE DIRECTIONS Addressing the mechanisms controlling GSH traffic in and out of the ER/periplasm and its recycling will help address GSH function in secretion. In addition, as thioredoxin reductase was recently implicated in ER oxidative protein folding, the relative contribution of each of these two reducing pathways should now be addressed. Antioxid. Redox Signal. 27, 1178-1199.
Collapse
Affiliation(s)
- Agnès Delaunay-Moisan
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| | - Alise Ponsero
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michel B Toledano
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
93
|
Abstract
SIGNIFICANCE Glutathione metabolism is comparable to a jigsaw puzzle with too many pieces. It is supposed to comprise (i) the reduction of disulfides, hydroperoxides, sulfenic acids, and nitrosothiols, (ii) the detoxification of aldehydes, xenobiotics, and heavy metals, and (iii) the synthesis of eicosanoids, steroids, and iron-sulfur clusters. In addition, glutathione affects oxidative protein folding and redox signaling. Here, I try to provide an overview on the relevance of glutathione-dependent pathways with an emphasis on quantitative data. Recent Advances: Intracellular redox measurements reveal that the cytosol, the nucleus, and mitochondria contain very little glutathione disulfide and that oxidative challenges are rapidly counterbalanced. Genetic approaches suggest that iron metabolism is the centerpiece of the glutathione puzzle in yeast. Furthermore, recent biochemical studies provide novel insights on glutathione transport processes and uncoupling mechanisms. CRITICAL ISSUES Which parts of the glutathione puzzle are most relevant? Does this explain the high intracellular concentrations of reduced glutathione? How can iron-sulfur cluster biogenesis, oxidative protein folding, or redox signaling occur at high glutathione concentrations? Answers to these questions not only seem to depend on the organism, cell type, and subcellular compartment but also on different ideologies among researchers. FUTURE DIRECTIONS A rational approach to compare the relevance of glutathione-dependent pathways is to combine genetic and quantitative kinetic data. However, there are still many missing pieces and too little is known about the compartment-specific repertoire and concentration of numerous metabolites, substrates, enzymes, and transporters as well as rate constants and enzyme kinetic patterns. Gathering this information might require the development of novel tools but is crucial to address potential kinetic competitions and to decipher uncoupling mechanisms to solve the glutathione puzzle. Antioxid. Redox Signal. 27, 1130-1161.
Collapse
Affiliation(s)
- Marcel Deponte
- Department of Parasitology, Ruprecht-Karls University , Heidelberg, Germany
| |
Collapse
|
94
|
Jain BP, Pandey S, Saleem N, Tanti GK, Mishra S, Goswami SK. SG2NA is a regulator of endoplasmic reticulum (ER) homeostasis as its depletion leads to ER stress. Cell Stress Chaperones 2017; 22:853-866. [PMID: 28634818 PMCID: PMC5655373 DOI: 10.1007/s12192-017-0816-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 01/24/2023] Open
Abstract
SG2NA belongs to a three-member striatin subfamily of WD40 repeat superfamily of proteins. It has multiple protein-protein interaction domains involved in assembling supramolecular signaling complexes. Earlier, we had demonstrated that there are at least five variants of SG2NA generated by alternative splicing, intron retention, and RNA editing. Such versatile and dynamic mode of regulation implicates it in tissue development. In order to shed light on its role in cell physiology, total proteome analysis was performed in NIH3T3 cells depleted of 78 kDa SG2NA, the only isoform expressing therein. A number of ER stress markers were among those modulated after knockdown of SG2NA. In cells treated with the ER stressors thapsigargin and tunicamycin, expression of SG2NA was increased at both mRNA and protein levels. The increased level of SG2NA was primarily in the mitochondria and the microsomes. A mouse injected with thapsigargin also had an increase in SG2NA in the liver but not in the brain. Cell cycle analysis suggested that while loss of SG2NA reduces the level of cyclin D1 and retains a population of cells in the G1 phase, concurrent ER stress facilitates their exit from G1 and traverse through subsequent phases with concomitant cell death. Thus, SG2NA is a component of intrinsic regulatory pathways that maintains ER homeostasis.
Collapse
Affiliation(s)
- Buddhi Prakash Jain
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University Bihar, Motihari, 845401, India
| | - Shweta Pandey
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Nikhat Saleem
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Goutam K Tanti
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
- Neuro-Kopf-Zentrum, Department of Neurology, Klinikumrechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str.22, 81675, Muenchen, Germany
| | - Shalini Mishra
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, New Delhi, -110054, India
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India.
| |
Collapse
|
95
|
Habash SS, Sobczak M, Siddique S, Voigt B, Elashry A, Grundler FMW. Identification and characterization of a putative protein disulfide isomerase (HsPDI) as an alleged effector of Heterodera schachtii. Sci Rep 2017; 7:13536. [PMID: 29051538 PMCID: PMC5648851 DOI: 10.1038/s41598-017-13418-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/21/2017] [Indexed: 12/27/2022] Open
Abstract
The plant-parasitic nematode Heterodera schachtii is an obligate biotroph that induces syncytial feeding sites in roots of its hosts. Nematodes produce effectors that are secreted into the host and facilitate infection process. Here we identified H. schachtii protein disulphide isomerase (HsPDI) as a putative effector that interferes with the host’s redox status. In situ hybridization showed that HsPdi is specifically localized within esophageal glands of pre-parasitic second stage juveniles (J2). HsPdi is up-regulated in the early parasitic J2s. Silencing of HsPdi by RNA interference in the J2s hampers their development and leads to structural malfunctions in associated feeding sites induced in Arabidopsis roots. Expression of HsPDI in Arabidopsis increases plant’s susceptibility towards H. schachtii. HsPdi expression is up-regulated in the presence of exogenous H2O2, whereas HsPdi silencing results in increased mortality under H2O2 stress. Stable expression of HsPDI in Arabidopsis plants decreases ROS burst induced by flg22. Transiently expressed HsPDI in N. benthamiana leaves is localized in the apoplast. HsPDI plays an important role in the interaction between nematode and plant, probably through inducing local changes in the redox status of infected host tissue. It also contributes to protect the nematode from exogenous H2O2 stress.
Collapse
Affiliation(s)
- Samer S Habash
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
| | - Miroslaw Sobczak
- Department of Botany, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, PL-02787, Warsaw, Poland
| | - Shahid Siddique
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany
| | - Boris Voigt
- Rheinische Friedrich-Wilhelms-University of Bonn, Department of Plant Cell Biology, Institute of Cellular and Molecular Botany, Kirschallee 1, D-53115, Bonn, Germany
| | - Abdelnaser Elashry
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany.,Strube Research GmbH & Co. KG, Hauptstraße 1, 38387, Söllingen, Germany
| | - Florian M W Grundler
- Rheinische Friedrich-Wilhelms-University of Bonn, INRES - Molecular Phytomedicine, Karlrobert-Kreiten-Straße 13, D-53115, Bonn, Germany.
| |
Collapse
|
96
|
'Something in the way she moves': The functional significance of flexibility in the multiple roles of protein disulfide isomerase (PDI). BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1383-1394. [PMID: 28844745 PMCID: PMC5654723 DOI: 10.1016/j.bbapap.2017.08.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/30/2017] [Accepted: 08/10/2017] [Indexed: 01/15/2023]
Abstract
Protein disulfide isomerase (PDI) has diverse functions in the endoplasmic reticulum as catalyst of redox transfer, disulfide isomerization and oxidative protein folding, as molecular chaperone and in multi-subunit complexes. It interacts with an extraordinarily wide range of substrate and partner proteins, but there is only limited structural information on these interactions. Extensive evidence on the flexibility of PDI in solution is not matched by any detailed picture of the scope of its motion. A new rapid method for simulating the motion of large proteins provides detailed molecular trajectories for PDI demonstrating extensive changes in the relative orientation of its four domains, great variation in the distances between key sites and internal motion within the core ligand-binding domain. The review shows that these simulations are consistent with experimental evidence and provide insight into the functional capabilities conferred by the extensive flexible motion of PDI.
Collapse
|
97
|
Verma P, Doharey PK, Yadav S, Omer A, Singh P, Saxena JK. Molecular cloning and characterization of protein disulfide isomerase of Brugia malayi, a human lymphatic filarial parasite. EXCLI JOURNAL 2017; 16:824-839. [PMID: 28827998 PMCID: PMC5547380 DOI: 10.17179/excli2017-214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/11/2017] [Indexed: 11/29/2022]
Abstract
Lymphatic filariasis results in an altered lymphatic system and the abnormal enlargement of body parts, causing pain, serious disability and social stigma. Effective vaccines are still not available nowadays, drugs against the disease is required. Protein disulfide isomerase (PDI) is an essential catalyst of the endoplasmic reticulum which is involved in folding and chaperone activities in different biological systems. Here, we report the enzymatic characterization of a Brugia malayi Protein disulfide isomerase (BmPDI), which was expressed and purified from Escherichia coli BL21 (DE3). Western blotting analysis showed the recombinant BmPDI could be recognized by anti-BmPDI Rabbit serum. The rBmPDI exhibited an optimum activity at pH 8 and 40 °C. The enzyme was inhibited by aurin and PDI inhibitor. Recombinant BmPDI showed interaction with recombinant Brugia malayi calreticulin (rBmCRT). The three-dimensional model for BmPDI and BmCRT was generated by homology modelling. A total of 25 hydrogen bonds were found to be formed between two interfaces. There are 259 non-bonded contacts present in the BmPDI-BmCRT complex and 12 salt bridges were formed in the interaction.
Collapse
Affiliation(s)
- Pravesh Verma
- Division of Biochemistry, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Pawan Kumar Doharey
- Division of Biochemistry, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Sunita Yadav
- Division of Biochemistry, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Ankur Omer
- Division of Toxicology, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Poonam Singh
- Division of Toxicology, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Jitendra Kumar Saxena
- Division of Biochemistry, CSIR-Central Drug Research Institute, BS10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|
98
|
Abstract
Cysteine thiols are among the most reactive functional groups in proteins, and their pairing in disulfide linkages is a common post-translational modification in proteins entering the secretory pathway. This modest amino acid alteration, the mere removal of a pair of hydrogen atoms from juxtaposed cysteine residues, contrasts with the substantial changes that characterize most other post-translational reactions. However, the wide variety of proteins that contain disulfides, the profound impact of cross-linking on the behavior of the protein polymer, the numerous and diverse players in intracellular pathways for disulfide formation, and the distinct biological settings in which disulfide bond formation can take place belie the simplicity of the process. Here we lay the groundwork for appreciating the mechanisms and consequences of disulfide bond formation in vivo by reviewing chemical principles underlying cysteine pairing and oxidation. We then show how enzymes tune redox-active cofactors and recruit oxidants to improve the specificity and efficiency of disulfide formation. Finally, we discuss disulfide bond formation in a cellular context and identify important principles that contribute to productive thiol oxidation in complex, crowded, dynamic environments.
Collapse
Affiliation(s)
- Deborah Fass
- Department of Structural Biology, Weizmann Institute of Science , Rehovot 7610001, Israel
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware 19716, United States
| |
Collapse
|
99
|
Gutiérrez T, Simmen T. Endoplasmic reticulum chaperones tweak the mitochondrial calcium rheostat to control metabolism and cell death. Cell Calcium 2017; 70:64-75. [PMID: 28619231 DOI: 10.1016/j.ceca.2017.05.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 12/16/2022]
Abstract
The folding of secretory proteins is a well-understood mechanism, based on decades of research on endoplasmic reticulum (ER) chaperones. These chaperones interact with newly imported polypeptides close to the ER translocon. Classic examples for these proteins include the immunoglobulin binding protein (BiP/GRP78), and the lectins calnexin and calreticulin. Although not considered chaperones per se, the ER oxidoreductases of the protein disulfide isomerase (PDI) family complete the folding job by catalyzing the formation of disulfide bonds through cysteine oxidation. Research from the past decade has demonstrated that ER chaperones are multifunctional proteins. The regulation of ER-mitochondria Ca2+ crosstalk is one of their additional functions, as shown for calnexin, BiP/GRP78 or the oxidoreductases Ero1α and TMX1. This function depends on interactions of this group of proteins with the ER Ca2+ handling machinery. This novel function makes perfect sense for two reasons: i. It allows ER chaperones to control mitochondrial apoptosis instantly without a lengthy bypass involving the upregulation of pro-apoptotic transcription factors via the unfolded protein response (UPR); and ii. It allows the ER protein folding machinery to fine-tune ATP import via controlling the speed of mitochondrial oxidative phosphorylation. Therefore, the role of ER chaperones in regulating ER-mitochondria Ca2+ flux identifies the progression of secretory protein folding as a central regulator of cell survival and death, at least in cell types that secrete large amount of proteins. In other cell types, ER protein folding might serve as a sentinel mechanism that monitors cellular well-being to control cell metabolism and apoptosis. The selenoprotein SEPN1 is a classic example for such a role. Through the control of ER-mitochondria Ca2+-flux, ER chaperones and folding assistants guide cellular apoptosis and mitochondrial metabolism.
Collapse
Affiliation(s)
- Tomas Gutiérrez
- Faculty of Medicine and Dentistry, Department of Cell Biology, University of Alberta, Edmonton, T6G2H7, Canada
| | - Thomas Simmen
- Faculty of Medicine and Dentistry, Department of Cell Biology, University of Alberta, Edmonton, T6G2H7, Canada,.
| |
Collapse
|
100
|
Maegawa KI, Watanabe S, Noi K, Okumura M, Amagai Y, Inoue M, Ushioda R, Nagata K, Ogura T, Inaba K. The Highly Dynamic Nature of ERdj5 Is Key to Efficient Elimination of Aberrant Protein Oligomers through ER-Associated Degradation. Structure 2017; 25:846-857.e4. [PMID: 28479060 DOI: 10.1016/j.str.2017.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/08/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
ERdj5, composed of an N-terminal J domain followed by six thioredoxin-like domains, is the largest protein disulfide isomerase family member and functions as an ER-localized disulfide reductase that enhances ER-associated degradation (ERAD). Our previous studies indicated that ERdj5 comprises two regions, the N- and C-terminal clusters, separated by a linker loop and with distinct functional roles in ERAD. We here present a new crystal structure of ERdj5 with a largely different cluster arrangement relative to that in the original crystal structure. Single-molecule observation by high-speed atomic force microscopy visualized rapid cluster movement around the flexible linker loop, indicating the highly dynamic nature of ERdj5 in solution. ERdj5 mutants with a fixed-cluster orientation compromised the ERAD enhancement activity, likely because of less-efficient reduction of aberrantly formed disulfide bonds and prevented substrate transfer in the ERdj5-mediated ERAD pathway. We propose a significant role of ERdj5 conformational dynamics in ERAD of disulfide-linked oligomers.
Collapse
Affiliation(s)
- Ken-Ichi Maegawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan; CREST, JST, Japan
| | - Satoshi Watanabe
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan; CREST, JST, Japan
| | - Kentaro Noi
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan; CREST, JST, Japan
| | - Masaki Okumura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan
| | - Yuta Amagai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan
| | - Michio Inoue
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan; CREST, JST, Japan
| | - Ryo Ushioda
- Department of Molecular Biosciences, Kyoto Sangyo University, Kyoto 603-8455, Japan; CREST, JST, Japan
| | - Kazuhiro Nagata
- Department of Molecular Biosciences, Kyoto Sangyo University, Kyoto 603-8455, Japan; CREST, JST, Japan
| | - Teru Ogura
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan; CREST, JST, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan; CREST, JST, Japan.
| |
Collapse
|